Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Nutr Biochem ; 123: 109512, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37907171

RESUMO

Long-term consumption of a high-fat diet (HFD) disrupts energy homeostasis and leads to weight gain. The fat mass and obesity-associated (FTO) gene has been consistently identified to be associated with HFD-induced obesity. The hypothalamus is crucial for regulating energy balance, and HFD-induced hypothalamic leptin resistance contributes to obesity. FTO, an N6-methyladenosine (m6A) RNA methylation regulator, may be a key mediator of leptin resistance. However, the exact mechanisms remain unclear. Therefore, the present study aims to investigate the association between FTO and leptin resistance. After HFD or standard diet (SD) feeding in male mice for 22 weeks, m6A-sequencing and western blotting assays were used to identify target genes and assess protein level, and molecular interaction changes. CRISPR/Cas9 gene knockout system was employed to investigate the potential function of FTO in leptin resistance and obesity. Our data showed that chemokine (C-X3-C motif) ligand 1 (CX3CL1) was a direct downstream target of FTO-mediated m6A modification. Furthermore, upregulation of FTO/CX3CL1 and suppressor of cytokine signaling 3 (SOCS3) in the hypothalamus impaired leptin-signal transducer and activator of transcription 3 signaling, resulting in leptin resistance and obesity. Compared to wild-type (WT) mice, FTO deficiency in leptin receptor-expressing neurons of the hypothalamus significantly inhibited the upregulation of CX3CL1 and SOCS3, and partially ameliorating leptin resistance under HFD conditions. Our findings reveal that FTO involved in the hypothalamic leptin resistance and provides novel insight into the function of FTO in the contribution to hypothalamic leptin resistance and obesity.


Assuntos
Dieta Hiperlipídica , Leptina , Animais , Masculino , Camundongos , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Quimiocina CX3CL1/metabolismo , Dieta Hiperlipídica/efeitos adversos , Hipotálamo/metabolismo , Leptina/metabolismo , Camundongos Endogâmicos C57BL , Obesidade/genética , Obesidade/metabolismo , Proteínas Supressoras da Sinalização de Citocina/genética
2.
J Tradit Chin Med ; 43(6): 1072-1080, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37946469

RESUMO

OBJECTIVE: To explore whether fat mass and obesity associated proteins (FTO) is an important target of Qiteng Xiaozhuo granules (QTXZG,) medicated serum in regulating proliferation and apoptosis of glomerular mesangial cells. METHODS: Medicated serum was obtained from Sprague-Dawley (SD) rats administered intragastrically with QTXZG decoction. The optimal concentration and intervention time of medicated serum were selected with the cell counting kit 8 assay. Cell proliferation was assessed by 5-ethynyl-2'-deoxyuridine (EdU) and cell apoptosis was investigated using flow cytometry. The expression of FTO, Proliferating cell nuclear antigen, Cyclin D1, B-cell lymphoma 2 (Bcl2) and BCL2 assaciated X was detected by Western blot and Real-time quantitative polymerase chain reaction, respectively. Quantification of the m6A RNA methylation was utilized to determine the total level of m6A methylation modification. RESULTS: EdU and flow cytometry assays revealed that QTXZG medicated serum can remarkably inhibit proliferation and promote apoptosis of lipopolysaccharide (LPS)-induced human glomerular mesangial cells (HGMCs). The FTO overexpression plasmid could inhibit proliferation and promote apoptosis of LPS-induced HGMCs. The FTO inhibitor (FB23-2) can significantly attenuate the effect of QTZXG medicated serum on inhibiting excessive proliferation and promoting apoptosis. QTXZG medicated serum can significantly increase FTO expression and decrease the level of m6A methylation modification. CONCLUSIONS: FTO is a key target for QTXZG medicated serum in inhibiting excessive proliferation and promoting apoptosis of human glomerular mesangial cells.


Assuntos
Lipopolissacarídeos , Células Mesangiais , Ratos , Animais , Humanos , Células Mesangiais/metabolismo , Ratos Sprague-Dawley , Lipopolissacarídeos/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proliferação de Células , Apoptose , Obesidade/tratamento farmacológico , Obesidade/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo
3.
J Nutr Biochem ; 122: 109437, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37666478

RESUMO

Obesity has become a major health crisis in the past decades. Branched-chain amino acids (BCAA), a class of essential amino acids, exerted beneficial health effects with regard to obesity and its related metabolic dysfunction, although the underlying reason is unknown. Here, we show that BCAA supplementation alleviates high-fat diet (HFD)-induced obesity and insulin resistance in mice and inhibits adipogenesis in 3T3-L1 cells. Further, we find that BCAA prevent the mitotic clonal expansion (MCE) of preadipocytes by reducing cyclin A2 (CCNA2) and cyclin-dependent kinase 2 (CDK2) expression. Mechanistically, BCAA decrease the concentration of nicotinamide adenine dinucleotide phosphate (NADPH) in adipose tissue and 3T3-L1 cells by reducing glucose-6-phosphate dehydrogenase (G6PD) expression. The reduced NADPH attenuates the expression of fat mass and obesity-associated (FTO) protein, a well-known m6A demethylase, to increase the N6-methyladenosine (m6A) levels of Ccna2 and Cdk2 mRNA. Meanwhile, the high m6A levels of Ccna2 and Cdk2 mRNA are recognized by YTH N6-methyladenosine RNA binding protein 2 (YTHDF2), which results in mRNA decay and reduction of their protein expressions. Overall, our data demonstrate that BCAA inhibit obesity and adipogenesis by reducing CDK2 and CCNA2 expression via an NADPH-FTO-m6A coordinated manner in vivo and in vitro, which raises a new perspective on the role of m6A in the BCAA regulation of obesity and adipogenesis.


Assuntos
Aminoácidos de Cadeia Ramificada , Obesidade , Camundongos , Animais , NADP , Aminoácidos de Cadeia Ramificada/metabolismo , Obesidade/metabolismo , Ciclo Celular , Adipogenia , RNA Mensageiro/metabolismo , Células 3T3-L1 , Dieta Hiperlipídica/efeitos adversos , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo
4.
Neuroendocrinology ; 113(1): 80-91, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36030776

RESUMO

INTRODUCTION: Fat mass and obesity-associated (FTO) gene is strongly associated with obesity which brings a major health threat. Altered expression of its encoded protein FTO in the hypothalamus has been identified to contribute to central control of appetite and body weight. However, its molecular mechanisms remain elusive. METHODS: Mouse hypothalamic POMC cell line N43/5 was treated with FTO inhibitor rhein, FTO shRNA, or extracellular signal-regulated kinase 1/2 (ERK1/2) inhibitor U0126 to inhibit FTO or ERK1/2. Rhein and U0126 were injected into lateral ventricle of the mice by intracerebroventricular cannulation. Western blotting and immunofluorescent assays were performed to monitor protein level. RESULTS: This study identified that inhibition of FTO in N43/5 cells led to phosphorylation of signal transducer and activator of transcription 3 (STAT3) at S727 site and induced p-STAT3-S727 nuclear translocation. We further showed that FTO inhibition promoted phosphorylation of ERK1/2; specific inhibition of ERK1/2 signaling by U0126 could abolish the effect of FTO inhibition on STAT3-S727 phosphorylation and nuclear translocation. Furthermore, we found that inhibition of hypothalamic FTO promoted STAT3-S727 phosphorylation in the hypothalamic arcuate nucleus, and the mice showed reductions in food intake and body weight. In addition, inhibition of hypothalamic ERK1/2 could abolish the effects of FTO inhibition on STAT3-S727 phosphorylation, reductions of food intake and body weight. CONCLUSION: Our in vitro and in vivo data suggest that the inhibition of hypothalamic FTO could activate STAT3 through ERK1/2, which is potentially associated with reductions in food intake and body weight.


Assuntos
Sistema de Sinalização das MAP Quinases , Fator de Transcrição STAT3 , Camundongos , Animais , Fator de Transcrição STAT3/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Hipotálamo/metabolismo , Peso Corporal , Obesidade/metabolismo , Ingestão de Alimentos , Fosforilação , Leptina/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo
5.
Ann Hepatol ; 28(1): 100874, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36371078

RESUMO

Obesity is a risk factor for developing nonalcoholic fatty liver disease (NAFLD), and the associated molecular mechanisms could be targeted with nutrient-based strategies. Therefore, it is necessary to review the current mechanisms to propose further treatments. Obesity facilitates the onset of insulin resistance, lipidic abnormalities, hepatic fat accumulation, lipid peroxidation, mitochondrial dysfunction, excessive reactive oxygen species (ROS) production, and inflammation, all related to further steatosis progression and fibrosis. Microbiota alterations can also influence liver disease by the translocation of pathogenic bacteria, energy extraction from short chain fatty acids (SCFAs), intestinal suppression of the expression of fasting-induced adipose factor (FIAF), reduction of bile acids, and altered choline metabolism. There are also genetic polymorphisms in metabolic proteins that predispose to a higher risk of liver diseases, such as those found in the patatin-like phospholipase domain-containing 3 (PNPLA3), transmembrane 6 superfamily member 2 (TM6SF2), membrane-bound O-acyltransferase domain-containing 7 (MBOAT7) or also known as lysophosphatidylinositol acyltransferase 1 (LPIAT1), transmembrane channel-like 4 genes (TMC4), fat mass and obesity-associated protein (FTO), the b Klotho (KLB) and carboxylesterase (CES1). No clear dietary guidelines target all mechanisms related to NAFLD development and progression. However, energy and carbohydrate intake restriction, regular physical exercise, supplementation of antioxidants, and restoration of gut microbiota seem to have beneficial effects on the new proposed features of NAFLD.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Humanos , Hepatopatia Gordurosa não Alcoólica/diagnóstico , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Fígado/patologia , Obesidade/genética , Obesidade/metabolismo , Fatores de Risco , Nutrientes , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo
6.
Int J Obes (Lond) ; 46(11): 2029-2039, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36115924

RESUMO

OBJECTIVE: Obesity, a leading cause of several metabolic abnormalities, is mainly caused by imbalanced energy homeostasis. IRX3 and IRX5 have been suggested as genetic determinants of obesity in connection with the intronic variants of the FTO gene, the strongest genetic risk factor of polygenic obesity in humans. Although the causal effects of Irx3 and its cooperation with Irx5 in obesity and associated metabolic abnormalities have been demonstrated in vivo, the function of Irx5 in energy homeostasis remains unclear. Here we aim to decipher the actions of Irx5 in the regulation of obesity and metabolic abnormalities. METHODS: We employed a mouse model homozygous for an Irx5-knockout (Irx5KO) allele and determined its metabolic phenotype in the presence or absence of a high-fat diet challenge. To investigate the function of Irx5 in the regulation of energy homeostasis, adipose thermogenesis and hypothalamic leptin response were assessed, and single-cell RNA sequencing (scRNA-seq) in the hypothalamic arcuate-median eminence (ARC-ME) was conducted. RESULTS: Irx5KO mice were leaner and resistant to diet-induced obesity as well as associated metabolic abnormalities, primarily through loss of adiposity. Assessments of energy expenditure and long-term dietary intake revealed that an increase in basal metabolic rate with adipose thermogenesis and a reduction of food intake with improved hypothalamic leptin response in Irx5KO mice may contribute to the anti-obesity effects. Utilizing scRNA-seq and marker gene analyses, we demonstrated the number of ARC-ME neurons was elevated in Irx5KO mice, suggesting a direct role for Irx5 in hypothalamic feeding control. CONCLUSIONS: Our study demonstrates that Irx5 is a genetic factor determining body mass/composition and obesity and regulates both energy expenditure and intake.


Assuntos
Leptina , Obesidade , Humanos , Animais , Camundongos , Leptina/metabolismo , Obesidade/genética , Obesidade/metabolismo , Dieta Hiperlipídica , Hipotálamo/metabolismo , Metabolismo Energético/genética , Camundongos Knockout , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo
7.
BMC Biol ; 20(1): 39, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35135551

RESUMO

BACKGROUND: Obesity leads to a decline in the exercise capacity of skeletal muscle, thereby reducing mobility and promoting obesity-associated health risks. Dietary intervention has been shown to be an important measure to regulate skeletal muscle function, and previous studies have demonstrated the beneficial effects of docosahexaenoic acid (DHA; 22:6 ω-3) on skeletal muscle function. At the molecular level, DHA and its metabolites were shown to be extensively involved in regulating epigenetic modifications, including DNA methylation, histone modifications, and small non-coding microRNAs. However, whether and how epigenetic modification of mRNA such as N6-methyladenosine (m6A) mediates DHA regulation of skeletal muscle function remains unknown. Here, we analyze the regulatory effect of DHA on skeletal muscle function and explore the involvement of m6A mRNA modifications in mediating such regulation. RESULTS: DHA supplement prevented HFD-induced decline in exercise capacity and conversion of muscle fiber types from slow to fast in mice. DHA-treated myoblasts display increased mitochondrial biogenesis, while slow muscle fiber formation was promoted through DHA-induced expression of PGC1α. Further analysis of the associated molecular mechanism revealed that DHA enhanced expression of the fat mass and obesity-associated gene (FTO), leading to reduced m6A levels of DNA damage-induced transcript 4 (Ddit4). Ddit4 mRNA with lower m6A marks could not be recognized and bound by the cytoplasmic m6A reader YTH domain family 2 (YTHDF2), thereby blocking the decay of Ddit4 mRNA. Accumulated Ddit4 mRNA levels accelerated its protein translation, and the consequential increased DDIT4 protein abundance promoted the expression of PGC1α, which finally elevated mitochondria biogenesis and slow muscle fiber formation. CONCLUSIONS: DHA promotes mitochondrial biogenesis and skeletal muscle fiber remodeling via FTO/m6A/DDIT4/PGC1α signaling, protecting against obesity-induced decline in skeletal muscle function.


Assuntos
Dioxigenase FTO Dependente de alfa-Cetoglutarato , Ácidos Docosa-Hexaenoicos , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Animais , Dieta , Ácidos Docosa-Hexaenoicos/metabolismo , Ácidos Docosa-Hexaenoicos/farmacologia , Camundongos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Obesidade , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/farmacologia , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo
8.
Mol Cell Endocrinol ; 542: 111523, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-34843901

RESUMO

The GABAA receptor (GABAAR) plays important roles in the regulation of Mn-induced GnRH secretion in immature female rats. However, the underlying molecular mechanisms remain unknown. Here, we assessed whether FTO and its substrate m6A are correlated with GABAAR expression in GnRH neurons after treatment with Mn in vitro and in vivo. Our study indicated that Mn treatment increased the expression of GnRH mRNA and decreased the levels of GABAAR protein but had no effect on GABAAR mRNA. Moreover, Mn upregulated the levels of FTO and inhibited global cellular m6A levels and GABAAα2 mRNA m6A levels. Knockdown of FTO increased the expression of GABAAR protein and GABAAα2 mRNA m6A levels. Data from rat models further demonstrate that inhibition of FTO suppressed GABAAR protein expression in the hypothalamus, causing delayed puberty onset. Collectively, our findings suggest that FTO-dependent m6A demethylation plays a critical role in regulating GABAAR mRNA processing in GnRH neurons.


Assuntos
Hormônio Liberador de Gonadotropina , Puberdade Precoce , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Animais , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Puberdade Precoce/induzido quimicamente , Puberdade Precoce/genética , Puberdade Precoce/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Maturidade Sexual
9.
Nat Biotechnol ; 39(12): 1581-1588, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34294912

RESUMO

RNA N6-methyladenosine (m6A) modifications are essential in plants. Here, we show that transgenic expression of the human RNA demethylase FTO in rice caused a more than threefold increase in grain yield under greenhouse conditions. In field trials, transgenic expression of FTO in rice and potato caused ~50% increases in yield and biomass. We demonstrate that the presence of FTO stimulates root meristem cell proliferation and tiller bud formation and promotes photosynthetic efficiency and drought tolerance but has no effect on mature cell size, shoot meristem cell proliferation, root diameter, plant height or ploidy. FTO mediates substantial m6A demethylation (around 7% of demethylation in poly(A) RNA and around 35% decrease of m6A in non-ribosomal nuclear RNA) in plant RNA, inducing chromatin openness and transcriptional activation. Therefore, modulation of plant RNA m6A methylation is a promising strategy to dramatically improve plant growth and crop yield.


Assuntos
Oryza , Solanum tuberosum , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Biomassa , Desmetilação , Humanos , Plantas Geneticamente Modificadas/genética , Plantas Geneticamente Modificadas/metabolismo , RNA de Plantas/genética , Solanum tuberosum/genética
10.
Semin Cell Dev Biol ; 110: 43-50, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32466971

RESUMO

An emerging number of rare genetic disorders termed ciliopathies are associated with pediatric obesity. It is becoming clear that the mechanisms associated with cilia dysfunction and obesity in these syndromes are complex. In addition to ciliopathic syndromic forms of obesity, several cilia-associated signaling gene mutations also lead to morbid obesity. While cilia have critical and diverse functions in energy homeostasis including their roles in centrally mediated food intake as well as in peripheral tissues, many questions remain. Here, we briefly discuss the syndromic ciliopathies and monoallelic cilia signaling gene mutations associated with obesity. We also describe potential ways cilia may be involved in common obesity. We discuss how neuronal cilia impact food intake potentially through leptin signaling and changes in ciliary G protein-coupled receptor (GPCR) signaling. We highlight several recent studies that have implicated the potential for cilia in peripheral tissues such as adipose and the pancreas to contribute to metabolic dysfunction. Then we discuss the potential for cilia to impact energy homeostasis through their roles in both development and adult tissue homeostasis. The studies discussed in this review highlight how a comprehensive understanding of the requirement of cilia for the regulation of diverse biological functions will contribute to our understanding of common forms of obesity.


Assuntos
Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Cílios/metabolismo , Ciliopatias/genética , Leptina/genética , Obesidade Mórbida/genética , Obesidade Infantil/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Adulto , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Animais , Criança , Cílios/patologia , Ciliopatias/metabolismo , Ciliopatias/patologia , Ingestão de Alimentos/genética , Regulação da Expressão Gênica , Humanos , Hipotálamo/metabolismo , Hipotálamo/patologia , Leptina/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Obesidade Mórbida/metabolismo , Obesidade Mórbida/patologia , Pâncreas/metabolismo , Pâncreas/patologia , Obesidade Infantil/metabolismo , Obesidade Infantil/patologia , Transdução de Sinais
11.
Trends Endocrinol Metab ; 31(5): 368-377, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32035736

RESUMO

Genome-wide association studies (GWASs) have identified SNPs of the fat mass and obesity (FTO) gene as the most important risk alleles for obesity. However, how the presence of risk alleles affect phenotype is still a matter of intense investigation. In 2014, a study revealed that long-range enhancers from the intronic regions of the FTO gene regulate iroquois-class homeobox protein (IRX)3 expression. IRX3 is expressed in hypothalamic pro-opiomelanocortin (POMC) neurons and changes in its expression levels affect body adiposity by modifying food intake and energy expenditure. These findings have placed IRX3 as a potential target for the treatment of obesity. Here, we review studies that evaluated the roles of IRX3 in development, neurogenesis, and body energy homeostasis.


Assuntos
Tecido Adiposo Marrom/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Metabolismo Energético/fisiologia , Proteínas de Homeodomínio/metabolismo , Hipotálamo/metabolismo , Obesidade/metabolismo , Pró-Opiomelanocortina/metabolismo , Fatores de Transcrição/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Animais , Metabolismo Energético/genética , Proteínas de Homeodomínio/genética , Humanos , Obesidade/genética , Fatores de Transcrição/genética
12.
J Physiol Sci ; 69(6): 981-991, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31728912

RESUMO

Hypoglycemia-associated autonomic failure (HAAF) is a serious complication of diabetes which is associated with the absence of physiological homeostatic counter-regulatory mechanisms that are controlled by the hypothalamus and sympathetic nervous system. Identification of biomarkers for early detection of HAAF requires an advanced understanding of molecular signature of hypoglycemia which is yet to be identified. The outcomes of the present study have shown that the viability and the apoptotic rate of the hypothalamic neurons (mHypoE-N39) were decreased significantly due to hypoglycemia in a dose-dependent fashion (p < 0.05). Although there are more than 1000 miRNAs differentially expressed in hypothalamus, only twelve miRNAs (miR-7a, miR-7b, miR-9, miR-29b, miR-29c, miR-30a, miR-30b, miR-30c, miR-101b-3p, miR-181a-5p, miR-378-3p and miR-873-5p) were correlated to two main hypothalamic regulatory proteins, FOS and FTO. Expression of these proteins was very sensitive to hypoglycemia. We demonstrated that hypoglycemia modulates the expression of hypothalamic miRNAs that are related to FOS and FTO.


Assuntos
Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Glucose/administração & dosagem , Hipotálamo/citologia , MicroRNAs/metabolismo , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , MicroRNAs/genética , Proteínas Proto-Oncogênicas c-fos/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
13.
J Mol Cell Biol ; 11(2): 118-132, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29771336

RESUMO

Fat mass and obesity-associated (FTO) protein is a ferrous ion (Fe2+)/2-oxoglutarate (2-OG)-dependent demethylase preferentially catalyzing m6A sites in RNA. The FTO gene is highly expressed in the hypothalamus with fluctuation in response to various nutritional conditions, which is believed to be involved in the control of whole body metabolism. However, the underlying mechanism in response to different nutritional cues remains poorly understood. Here we show that ketogenic diet-derived ketone body ß-hydroxybutyrate (BHB) transiently increases FTO expression in both mouse hypothalamus and cultured cells. Interestingly, the FTO protein represses Fto promoter activity, which can be offset by BHB. We then demonstrate that FTO binds to its own gene promoter, and Fe2+, but not 2-OG, impedes this binding and increases FTO expression. The BHB-induced occupancy of the promoter by FTO influences the assembly of the basal transcriptional machinery. Importantly, a loss-of-function FTO mutant (I367F), which induces a lean phenotype in FTOI367F mice, exhibits augmented binding and elevated potency to repress the promoter. Furthermore, FTO fails to bind to its own promoter that promotes FTO expression in the hypothalamus of high-fat diet-induced obese and 48-h fasting mice, suggesting a disruption of the stable expression of this gene. Taken together, this study uncovers a new function of FTO as a Fe2+-sensitive transcriptional repressor dictating its own gene switch to form an auto-regulatory loop that may link with the hypothalamic control of body weight.


Assuntos
Dioxigenase FTO Dependente de alfa-Cetoglutarato , Peso Corporal/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Animais , Linhagem Celular , Regulação da Expressão Gênica , Hipotálamo/metabolismo , Camundongos , Células NIH 3T3 , Obesidade , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
14.
Nutr Neurosci ; 22(4): 243-252, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28914170

RESUMO

OBJECTIVE: The aim of the present study is to investigate the effects of topiramate on the fat mass/obesity-associated protein (FTO) and on the neuropeptide Y (NPY) level in the hypothalamus depending on the recently increased prevalence of obesity. METHOD: In this study, twenty-four female rats were divided into four equal groups: Non-obese control, obese control, non-obese topiramate, and obese topiramate. Obese groups were fed with a 40% high-fat diet. At the end of the 9th week, the drug treatment started and the subjects were treated with topiramate once a day for 6 weeks. All animals underwent cardiac perfusion under high-dose anesthesia on the 15th week. Tissues were analyzed using biochemical, histological, and stereological methods. RESULTS: In terms of neuron number in the arcuate nucleus area, a significant difference was observed among all groups (P < 0.01). The neuron number of the non-obese topiramate group was found to be significantly higher than that of the non-obese control group (P < 0.01). In the examination of the ventromedial nucleus of the entire group, it was observed that the neuron number of the non-obese control group was significantly lower than those of the other groups (P < 0.01). A significant increase in the NPY levels of the obese groups compared to the groups treated with topiramate was observed. Furthermore, the amount of the FTO protein increased in obese rats, while FTO and NPY levels decreased in the groups treated with topiramate. DISCUSSION: In conclusion, the mechanism of the effect of topiramate to create a state of obesity is thought to involve the decrease in the levels of NPY and FTO.


Assuntos
Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Fármacos Antiobesidade/administração & dosagem , Hipotálamo/efeitos dos fármacos , Neuropeptídeo Y/metabolismo , Obesidade/metabolismo , Topiramato/administração & dosagem , Animais , Feminino , Hipotálamo/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Obesidade/prevenção & controle , Ratos
15.
Nutrients ; 10(10)2018 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-30241328

RESUMO

Maternal diet and gestational hyperglycaemia have implications for offspring health. Leptin (LEP) and fat mass and obesity-associated (FTO) alleles are known to influence body fat mass in humans, potentially via effects on appetite. We hypothesized that expression of Fto, Lep, and other appetite-related genes (Argp, Npy, Pomc, Cart, Lepr) in the offspring of female mice are influenced by the glycaemic index (GI) of carbohydrates in the maternal diet. C57BL/6 mice were randomly assigned to low or high GI diets and mated with chow-fed males at eight weeks of age. Male pups were weaned at four weeks and randomly divided into two groups, one group following their mother's diet (LL and HH), and one following the standard chow diet (LC and HC) to 20 weeks. Fto expression was 3.8-fold higher in the placenta of mothers fed the high GI diet (p = 0.0001) and 2.5-fold higher in the hypothalamus of 20-week old offspring fed the high GI (HH vs. LL, p < 0.0001). By contrast, leptin gene (Lep) expression in visceral adipose tissue was 4.4-fold higher in four-week old offspring of low GI mothers (LC vs. HC, p < 0.0001) and 3.3-fold higher in visceral adipose tissue of 20-week old animals (LL vs. HH, p < 0.0001). Plasma ghrelin and leptin levels, and hypothalamic appetite genes were also differentially regulated by maternal and offspring diet. These findings provide the first evidence in an animal model that maternal high GI dietary carbohydrates that are digested and absorbed faster may contribute to programming of appetite in offspring.


Assuntos
Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Carboidratos da Dieta/administração & dosagem , Índice Glicêmico , Leptina/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Estado Nutricional , Efeitos Tardios da Exposição Pré-Natal , Proteína Relacionada com Agouti/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Animais , Carboidratos da Dieta/metabolismo , Feminino , Regulação da Expressão Gênica , Hipotálamo/metabolismo , Gordura Intra-Abdominal/metabolismo , Leptina/genética , Masculino , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeo Y/metabolismo , Placenta/metabolismo , Gravidez , Pró-Opiomelanocortina/metabolismo , Receptores para Leptina/metabolismo
16.
Int J Obes (Lond) ; 42(7): 1378-1388, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29795461

RESUMO

BACKGROUND/OBJECTIVE: N6-methyladenosine (m6A) modification of mRNA plays a role in regulating adipogenesis. However, its underlying mechanism remains largely unknown. Epigallocatechin gallate (EGCG), the most abundant catechin in green tea, plays a critical role in anti-obesity and anti-adipogenesis. METHODS: High-performance liquid chromatography coupled with triple-quadrupole tandem mass spectrometry (HPLC-QqQ-MS/MS) was performed to determine the m6A levels in 3T3-L1 preadipocytes. The effects of EGCG on the m6A levels in specific genes were determined by methylated RNA immunoprecipitation coupled with quantitative real-time PCR (meRIP-qPCR). Several adipogenesis makers and cell cycle genes were analyzed by quantitative real-time PCR (qPCR) and western blotting. Lipid accumulation was evaluated by oil red O staining. All measurements were performed at least for three times. RESULTS: Here we showed that EGCG inhibited adipogenesis by blocking the mitotic clonal expansion (MCE) at the early stage of adipocyte differentiation. Exposing 3T3-L1 cells to EGCG reduced the expression of fat mass and obesity-associated (FTO) protein, an m6A demethylase, which led to increased overall levels of RNA m6A methylation. Cyclin A2 (CCNA2) and cyclin dependent kinase 2 (CDK2) play vital roles in MCE. The m6A levels of CCNA2 and CDK2 mRNA were dramatically enhanced by EGCG. Interestingly, EGCG increased the expression of YTH N6-methyladenosine RNA binding protein 2 (YTHDF2), which recognized and decayed methylated mRNAs, resulting in decreased protein levels of CCNA2 and CDK2. As a result, MCE was blocked and adipogenesis was inhibited. FTO overexpression and YTHDF2 knockdown in 3T3-L1 cells significantly increased CCNA2 and CDK2 protein levels and ameliorated the EGCG-induced adipogenesis inhibition. Thus, m6A-dependent CCNA2 and CDK2 expressions mediated by FTO and YTHDF2 contributed to EGCG-induced adipogenesis inhibition. CONCLUSION: Our findings provide mechanistic insights into how m6A is involved in the EGCG regulation of adipogenesis and shed light on its anti-obesity effect.


Assuntos
Adipócitos/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Fármacos Antiobesidade/farmacologia , Catequina/análogos & derivados , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Células 3T3-L1/citologia , Adipócitos/citologia , Dioxigenase FTO Dependente de alfa-Cetoglutarato/deficiência , Animais , Catequina/farmacologia , Modelos Animais de Doenças , Camundongos , RNA Mensageiro/química , RNA Mensageiro/genética , Chá/química
17.
Phytother Res ; 32(1): 84-93, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29193419

RESUMO

The metabolic syndrome (MetS) is a multicomponent condition with a complex etiology involving genetic and environmental factors. Artichoke leaf extract (ALE) has shown favorable effects on lipid and glucose metabolism. The present study aimed to investigate the effects of ALE supplementation on metabolic parameters in women with MetS, using a nutrigenetics approach. In this double-blind randomized clinical trial, 50 women (aged 20-50 years) with MetS were randomly allocated into the two groups: "ALE group" (received 1,800 mg hydroalcoholic extract of artichoke as four tablets per day) and "placebo group" (received placebo consisted of corn starch, lactose, and avicel as four tablets per day) for 12 weeks. The biochemical and anthropometric parameters were determined before and after the intervention. The FTO-rs9939609 and the TCF7L2-rs7903146 polymorphisms were genotyped by polymerase chain reaction-restriction fragment length polymorphism. In carriers of A allele of the FTO-rs9939609, ALE supplementation resulted in a statistically significant decrease in serum triglyceride level compared with placebo (-19.11% vs. 10.83%; p < .05), with no other significant differences between the two groups. The TCF7L2-rs7903146 polymorphism showed no interaction with response to ALE (p > .05). These findings suggest that ALE supplementation may improve serum triglyceride level in A allele genotype of FTO-rs9939609 polymorphism in women with MetS.


Assuntos
Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Cynara scolymus/química , Síndrome Metabólica/tratamento farmacológico , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Adulto , Método Duplo-Cego , Feminino , Genótipo , Humanos , Masculino , Síndrome Metabólica/patologia , Pessoa de Meia-Idade , Polimorfismo Genético , Adulto Jovem
18.
J Clin Invest ; 126(5): 1897-910, 2016 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-27064284

RESUMO

Noncoding polymorphisms in the fat mass and obesity-associated (FTO) gene represent common alleles that are strongly associated with effects on food intake and adiposity in humans. Previous studies have suggested that the obesity-risk allele rs8050136 in the first intron of FTO alters a regulatory element recognized by the transcription factor CUX1, thereby leading to decreased expression of FTO and retinitis pigmentosa GTPase regulator-interacting protein-1 like (RPGRIP1L). Here, we evaluated the effects of rs8050136 and another potential CUX1 element in rs1421085 on expression of nearby genes in human induced pluripotent stem cell-derived (iPSC-derived) neurons. There were allele-dosage effects on FTO, RPGRIP1L, and AKT-interacting protein (AKTIP) expression, but expression of other vicinal genes, including IRX3, IRX5, and RBL2, which have been implicated in mediating functional effects, was not altered. In vivo manipulation of CUX1, Fto, and/or Rpgrip1l expression in mice affected adiposity in a manner that was consistent with CUX1 influence on adiposity via remote effects on Fto and Rpgrip1l expression. In support of a mechanism, mice hypomorphic for Rpgrip1l exhibited hyperphagic obesity, as the result of diminished leptin sensitivity in Leprb-expressing neurons. Together, the results of this study indicate that the effects of FTO-associated SNPs on energy homeostasis are due in part to the effects of these genetic variations on hypothalamic FTO, RPGRIP1L, and possibly other genes.


Assuntos
Alelos , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Regulação da Expressão Gênica , Íntrons , Obesidade , Polimorfismo de Nucleotídeo Único , Proteínas , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Animais , Proteínas do Citoesqueleto , Metabolismo Energético/genética , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Humanos , Hipotálamo/metabolismo , Hipotálamo/patologia , Camundongos , Camundongos Transgênicos , Obesidade/genética , Obesidade/metabolismo , Obesidade/patologia , Proteínas/genética , Proteínas/metabolismo , Proteína p130 Retinoblastoma-Like/biossíntese , Proteína p130 Retinoblastoma-Like/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA